Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Free Radic Biol Med ; 143: 275-287, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31442556

RESUMO

Platelet-activating factor (PAF) is a potent inflammatory agonist. In Swiss albino mice, intraperitoneal injection of PAF causes sudden death with oxidative stress and disseminated intravascular coagulation (DIC), characterized by prolonged prothrombin time, thrombocytopenia, reduced fibrinogen content, and increased levels of fibrinogen degradation products. However, the underlying mechanism(s) is unknown. The PAF-R antagonist WEB-2086 protected mice against PAF-induced death by reducing DIC and oxidative stress. Accordingly, general antioxidants such as ascorbic acid, α-tocopherol, gallic acid, and N-acetylcysteine partially protected mice from PAF-induced death. N-acetylcysteine, a clinically used antioxidant, prevented death in 67% of mice, ameliorated DIC characteristics and histological alterations in the liver, and reduced oxidative stress. WEB-2086 suppressed H2O2-mediated oxidative stress in isolated mouse peritoneal macrophages, suggesting that PAF signaling may be a downstream effector of reactive oxygen species generation. PAF stimulated all three (ERK, JNK, and p38) of the MAP-kinases, which were also inhibited by N-acetylcysteine. Furthermore, a JNK inhibitor (SP600125) and ERK inhibitor (SCH772984) partially protected mice against PAF-induced death, whereas a p38 MAP-kinase inhibitor (SB203580) provided complete protection against DIC and death. In human platelets, which have the canonical PAF-R and functional MAP-kinases, JNK and p38 inhibitors abolished PAF-induced platelet aggregation, but the ERK inhibitor was ineffective. Our studies identify p38 MAP-kinase as a critical, but unrecognized component in PAF-induced mortality in mice. These findings suggest an alternative therapeutic strategy to address PAF-mediated pathogenicity, which plays a role in a broad range of inflammatory diseases.


Assuntos
Morte Súbita/prevenção & controle , Inibidores Enzimáticos/farmacologia , Estresse Oxidativo , Fator de Ativação de Plaquetas/toxicidade , Substâncias Protetoras/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Acetilcisteína/farmacologia , Animais , Morte Súbita/etiologia , Morte Súbita/patologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo
2.
PLoS One ; 11(4): e0153282, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27064683

RESUMO

Lipopolysaccharide (LPS) signaling through Toll-like receptor-4 (TLR-4) has been implicated in the pathogenesis of many infectious diseases. Some believe that TLR-mediated pathogenicity is due, in part, to the lipid pro-inflammatory mediator platelet-activating factor (PAF), but this has been questioned. To test the direct contribution of PAF in endotoxemia in murine models, we injected PAF intraperitoneally into Swiss albino mice in the presence and absence of LPS. PAF alone (5 µg/mouse) caused death within 15-20 min, but this could be prevented by pretreating mice with PAF-receptor (PAF-R) antagonists or PAF-acetylhydrolase (PAF-AH). A low dose of LPS (5 mg/kg body wt) did not impair PAF-induced death, whereas higher doses (10 or 20 mg/kg body wt) delayed death, probably via LPS cross-tolerance. Cross-tolerance occurred only when PAF was injected simultaneously with LPS or within 30 min of LPS injection. Tolerance does not appear to be due to an abundant soluble mediator. Histologic examination of lungs and liver and measurement of circulating TNF-α and IL-10 levels suggested that the inflammatory response is not diminished during cross-tolerance. Interestingly, aspirin, a non-specific cyclooxygenase (COX) inhibitor, partially blocked PAF-induced sudden death, whereas NS-398, a specific COX-2 inhibitor, completely protected mice from the lethal effects of PAF. Both COX inhibitors (at 20 mg/kg body wt) independently amplified the cross-tolerance exerted by higher dose of LPS, suggesting that COX-derived eicosanoids may be involved in these events. Thus, PAF does not seem to have a protective role in endotoxemia, but its effects are delayed by LPS in a COX-sensitive way. These findings are likely to shed light on basic aspects of the endotoxin cross-tolerance occurring in many disease conditions and may offer new opportunities for clinical intervention.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Morte Súbita/prevenção & controle , Endotoxemia/prevenção & controle , Lipopolissacarídeos/farmacologia , Fator de Ativação de Plaquetas/toxicidade , Prostaglandina-Endoperóxido Sintases/química , Animais , Citocinas/metabolismo , Morte Súbita/etiologia , Morte Súbita/patologia , Endotoxemia/etiologia , Endotoxemia/mortalidade , Endotoxemia/patologia , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fator de Ativação de Plaquetas/administração & dosagem , Glicoproteínas da Membrana de Plaquetas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Substâncias Protetoras/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Taxa de Sobrevida
3.
PLoS Genet ; 7(2): e1001299, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21347278

RESUMO

During Alzheimer's Disease, sustained exposure to amyloid-ß42 oligomers perturbs metabolism of ether-linked glycerophospholipids defined by a saturated 16 carbon chain at the sn-1 position. The intraneuronal accumulation of 1-O-hexadecyl-2-acetyl-sn-glycerophosphocholine (C16:0 PAF), but not its immediate precursor 1-O-hexadecyl-sn-glycerophosphocholine (C16:0 lyso-PAF), participates in signaling tau hyperphosphorylation and compromises neuronal viability. As C16:0 PAF is a naturally occurring lipid involved in cellular signaling, it is likely that mechanisms exist to protect cells against its toxic effects. Here, we utilized a chemical genomic approach to identify key processes specific for regulating the sensitivity of Saccharomyces cerevisiae to alkyacylglycerophosphocholines elevated in Alzheimer's Disease. We identified ten deletion mutants that were hypersensitive to C16:0 PAF and five deletion mutants that were hypersensitive to C16:0 lyso-PAF. Deletion of YDL133w, a previously uncharacterized gene which we have renamed SRF1 (Spo14 Regulatory Factor 1), resulted in the greatest differential sensitivity to C16:0 PAF over C16:0 lyso-PAF. We demonstrate that Srf1 physically interacts with Spo14, yeast phospholipase D (PLD), and is essential for PLD catalytic activity in mitotic cells. Though C16:0 PAF treatment does not impact hydrolysis of phosphatidylcholine in yeast, C16:0 PAF does promote delocalization of GFP-Spo14 and phosphatidic acid from the cell periphery. Furthermore, we demonstrate that, similar to yeast cells, PLD activity is required to protect mammalian neural cells from C16:0 PAF. Together, these findings implicate PLD as a potential neuroprotective target capable of ameliorating disruptions in lipid metabolism in response to accumulating oligomeric amyloid-ß42.


Assuntos
Glicerofosfolipídeos/metabolismo , Fosfolipase D/metabolismo , Fator de Ativação de Plaquetas/análogos & derivados , Fator de Ativação de Plaquetas/toxicidade , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Doença de Alzheimer/metabolismo , Linhagem Celular , Glicerofosfolipídeos/genética , Humanos , Metabolismo dos Lipídeos/genética , Mutação/genética , Neurônios/metabolismo , Fosfolipase D/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
4.
Neurochem Int ; 56(8): 893-905, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20362023

RESUMO

Calcium (Ca2+) dyshomeostasis may be of pivotal importance in mediating the neurotoxic action of amyloid beta peptide (Abeta), but the mechanism whereby Abeta disrupts Ca2+ homeostasis remains unclear. Using hippocampal neuronal cultures, the present study investigated possible mechanisms underlying Ca2+ dyshomeostasis induced by the oligomeric form of Abeta1-42 and two possible mediators of its toxicity, hydrogen peroxide (H2O2) and platelet-activating factor (PAF). It was found that, both H2O2 and PAF were able to reproduce each of the events induced by oligomeric Abeta1-42, including (a) Ca2+ influx via N-methyl-D-aspartic acid (NMDA) receptors, (b) enhancement of Ca2+ response to NMDA via activation of protein kinase C (PKC), (c) the increase of extracellular concentrations of glutamate and (d) the increase in cytosolic free Ca2+ ([Ca2+]i). Moreover, each of these events could be blocked by Ginkgo biloba extract EGb761, a free radical scavenger with PAF antagonism, and by quercetin, a constituent with well-established free radical scavenging property. In contrast, ginkgolide B, another constituent of EGb761 with well-established PAF-antagonizing activity protected the neurons against Ca2+ dyshomeostasis induced by Abeta1-42 and PAF, but not by H2O2. These results suggested the possibility that Abeta1-42-induced Ca2+ dyshomeostasis might be mediated by formation of toxic mediators such as H2O2 and PAF. Therefore, increased production of toxic mediators such as H2O2 and PAF in the brain may be critical in the pathological mechanism of neurodegenerative diseases, particularly Alzheimer's disease (AD), and may serve as major therapeutic targets for these diseases.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Cálcio/antagonistas & inibidores , Homeostase/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Fragmentos de Peptídeos/toxicidade , Extratos Vegetais/farmacologia , Fator de Ativação de Plaquetas/toxicidade , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Células Cultivadas , Ginkgo biloba/fisiologia , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Hipocampo/metabolismo , Homeostase/fisiologia , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Fragmentos de Peptídeos/antagonistas & inibidores , Extratos Vegetais/uso terapêutico , Fator de Ativação de Plaquetas/antagonistas & inibidores , Fator de Ativação de Plaquetas/metabolismo , Ratos , Ratos Sprague-Dawley
5.
Biochem Biophys Res Commun ; 385(4): 576-80, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19486886

RESUMO

Platelet activating factor (PAF), an endogenous bioactive phospholipid, has been documented as a pivotal mediator in the inflammatory cascade underlying the pathogenesis of many diseases including necrotizing enterocolitis. Much effort has been directed towards finding an effective in vivo inhibitor of PAF signaling. Here, we report that a small, highly stable, lysosomal lipid transport protein, the GM2 activator protein (GM2AP) is able to inhibit the inflammatory processes otherwise initiated by PAF in a rat model of necrotizing enterocolitis. Based on behavioral observations, gross anatomical observations at necropsy, histopathology and immunocytochemistry, the administration of recombinant GM2AP inhibits the devastating gastrointestinal necrosis resulting from the injection of rats with LPS and PAF. Recombinant GM2AP treatment not only markedly decrease tissue destruction, but also helped to maintain tight junction integrity at the gastrointestinal level as judged by contiguous Zonula Occludens-1 staining of the epithelial layer lining the crypts.


Assuntos
Enterocolite Necrosante/tratamento farmacológico , Proteína Ativadora de G(M2)/administração & dosagem , Fator de Ativação de Plaquetas/antagonistas & inibidores , Proteínas Recombinantes/administração & dosagem , Animais , Modelos Animais de Doenças , Enterocolite Necrosante/induzido quimicamente , Enterocolite Necrosante/etiologia , Enterocolite Necrosante/patologia , Masculino , Fator de Ativação de Plaquetas/metabolismo , Fator de Ativação de Plaquetas/toxicidade , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
6.
Biochem Pharmacol ; 77(7): 1223-35, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19283893

RESUMO

Platelet-activating factor (PAF) is an inflammatory mediator widely known to exert relevant pathophysiological functions. However, the relevance of PAF in nociception has received much less attention. Herein, we have investigated the mechanisms underlying PAF-induced spontaneous nociception and mechanical hypersensitivity in the rat paw. PAF injection (1- 30 nmol/paw) resulted in a dose-related overt nociception, whilst only the dose of 10 nmol/ paw produced a significant and time-related mechanical hypersensitivity. Local coinjection of PAF antagonist WEB2086 significantly inhibited both spontaneous nociception and mechanical hypersensitivity. Moreover, the coinjection of the natural IL-1beta receptor antagonist (IRA) notably prevented both PAF-induced nociceptive responses, whilst these responses were not altered by anti-TNFalpha coinjection. Interestingly, pretreatment with the ultrapotent vaniloid agonist resiniferotoxin, coinjection of the TRPV1 receptor antagonist SB366791, or mast cell depletion with compound 48/80 markedly prevented PAF-induced spontaneous nociception. Conversely, PAF-elicited mechanical hypersensitivity was strikingly susceptible to distinct antineutrophil-related strategies, namely the antineutrophil antibody, the selectin blocker fucoidin, the chemokine CXCR2 receptor antagonist SB225002, and the C5a receptor antibody anti-CD88. Notably, the same antineutrophil migration strategies significantly prevented the increase of myeloperoxidase activity induced by PAF. The mechanical hypersensitivity caused by PAF was also prevented by the cyclooxygenase inhibitors indomethacin or celecoxib, and by the selective beta(1) adrenergic receptor antagonist atenolol. Collectively, the present results provide consistent evidence indicating that distinct mechanisms are involved in the spontaneous nociception and mechanical hypersensitivity caused by PAF. They also support the concept that selective PAF receptor antagonists might constitute interesting targets for the development of new analgesic drugs.


Assuntos
Membro Anterior/metabolismo , Medição da Dor/métodos , Dor/induzido quimicamente , Dor/metabolismo , Fator de Ativação de Plaquetas/toxicidade , Animais , Azepinas/farmacologia , Azepinas/uso terapêutico , Membro Anterior/efeitos dos fármacos , Membro Anterior/patologia , Masculino , Dor/prevenção & controle , Medição da Dor/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Glicoproteínas da Membrana de Plaquetas/agonistas , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Triazóis/farmacologia , Triazóis/uso terapêutico
7.
J Lipid Res ; 49(10): 2250-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18550892

RESUMO

The platelet-activating factor (PAF) family of glycerophospholipids accumulates in damaged brain tissue following injury. Little is known about the role of individual isoforms in regulating neuronal survival. Here, we compared the neurotoxic and neuroprotective activities of 1-O-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine (C16-PAF) and 1-O-octadecyl-2-acetyl-sn-glycero-3-phosphocholine (C18-PAF) in cerebellar granule neurons. We find that both C16-PAF and C18-PAF cause PAF receptor-independent death but signal through different pathways. C16-PAF activates caspase-7, whereas C18-PAF triggers caspase-independent death in PAF receptor-deficient neurons. We further show that PAF receptor signaling is either pro- or anti-apoptotic, depending upon the identity of the sn-1 fatty acid of the PAF ligand. Activation of the PAF G-protein-coupled receptor (PAFR) by C16-PAF stimulation is anti-apoptotic and inhibits caspase-dependent death. Activation of PAFR by C18-PAF is pro-apoptotic. These results demonstrate the importance of the long-chain sn-1 fatty acid in regulating PAF-induced caspase-dependent apoptosis, caspase-independent neurodegeneration, and neuroprotection in the presence or absence of the PAF receptor.


Assuntos
Apoptose/efeitos dos fármacos , Carbono , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fator de Ativação de Plaquetas/química , Fator de Ativação de Plaquetas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Animais , Caspases/metabolismo , Linhagem Celular , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Micelas , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/química , Neurotoxinas/toxicidade , Éteres Fosfolipídicos/farmacologia , Fator de Ativação de Plaquetas/análogos & derivados , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/deficiência , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/metabolismo
8.
Cell Mol Neurobiol ; 28(1): 125-36, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17712622

RESUMO

(1) HIV-1 and viral proteins-evoked chronic brain inflammation, which is characterized by microglial activation, is the pivotal neuropathogenesis of HIV-1-associated dementia (HAD). Platelet-activating factor (PAF), mainly released from activated microglia and acts as a high potent inflammatory mediator and a neurotoxin, is indicated to be a principle initiator of neuroinflammation, neuronal dysfunction, and apoptosis related to HAD. Thus, bis-interacting ligands of acetylcholinesterase (AChE) inhibition and PAF receptor antagonism would be of great interest in the therapeutic potential of HAD not only for improvement of cognitive performance, but also for disease-modifying. (2). We have previously reported that a novel tetrahydrofuran-derived bis-interacting ligand PMS777 had satisfying potencies for PAF receptor blockade and AChE inhibition, and markedly improved cholinergic dysfunction-induced cognitive impairment in mice. Continuing with our research, we further investigated the neuroprotective activities of PMS777 on PAF-triggered neuronal injury in human neuroblastoma SH-SY5Y cells. (3) The bis-interacting ligand PMS777 (10 muM) obviously alleviated PAF-induced cell apoptosis in SH-SY5Y cells. Pretreatment with PMS777 also markedly inhibited intracellular Ca(2+) overload, down-regulation of anti-apoptotic bcl-2 mRNA, stimulation of pro-apoptotic bax mRNA expression and activation of caspase-3 pathway. Also, PMS777 could fine-tune pro-inflammatory cyclooxygenase-2 (cox-2) mRNA expression in PAF-treated cells. (4) These results suggest that PMS777 possesses a neuroprotective profile via anti-apoptotic/inflammatory signaling and warrant further investigations in connection with the potential value of this compound in HAD treatment.


Assuntos
Acetilcolinesterase/metabolismo , Apoptose/efeitos dos fármacos , Inibidores da Colinesterase/farmacologia , Furanos/farmacologia , Neurônios/patologia , Fator de Ativação de Plaquetas/análogos & derivados , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Cálcio/metabolismo , Inibidores de Caspase , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Ligantes , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Fator de Ativação de Plaquetas/toxicidade , Inibidores de Proteases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
9.
J Neuroimmune Pharmacol ; 2(2): 194-201, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18040844

RESUMO

The phospholipid mediator platelet-activating factor (PAF), an endogenous modulator of glutamatergic neurotransmission, can also be secreted by brain mononuclear phagocytes during HIV-1 infection. Platelet-activating factor can induce neuronal apoptosis by NMDA receptor-dependent and independent mechanisms. We now demonstrate that acute administration of sublethal doses of PAF to striatal slices augments synaptic facilitation in striatal neurons following high-frequency stimulation, which can be blocked by PAF receptor antagonists, suggesting that striatal synaptic facilitation can be augmented by PAF receptor agonism. We also demonstrate that repeated sublethal doses of PAF during tetanic stimulation can greatly increase the magnitude of postsynaptic potentials and action potentials, but a lethal dose of PAF destroys the capacity of corticostriatal synapses to achieve this augmented synaptic facilitation. Thus, the relative concentration and temporal pattern of PAF expression at glutamatergic synapses may govern whether it acts in a physiologic or pathophysiologic manner during striatal neurotransmission.


Assuntos
Corpo Estriado/fisiologia , Fosfolipídeos/fisiologia , Fator de Ativação de Plaquetas/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Corpo Estriado/efeitos dos fármacos , Relação Dose-Resposta a Droga , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipídeos/toxicidade , Fator de Ativação de Plaquetas/agonistas , Fator de Ativação de Plaquetas/toxicidade , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
11.
J Neurochem ; 103(1): 88-97, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17877634

RESUMO

The bioactive lipid mediator platelet activating factor (PAF) is recognized as a key effecter of neuronal apoptosis, yet it is not clear whether its G-protein coupled receptor (PAFR) initiates or prevents PAF neurotoxicity. Using PAFR-/- and congenic wild-type mice, we show that PAF triggers caspase-3/7 activity and neuronal death in PAFR-/- but not PAFR+/+ cerebellar granule neurons. Restoring receptor expression by recombinant adenoviral infection protected cells from PAF challenge. Neuronal death was not mediated by nitric oxide or N-methyl-d-aspartate receptor signaling given that N-nitro-l-arginine methyl ester and MK-801 did not inhibit PAF-induced neuronal loss in PAFR-/- neurons. To intervene in PAFR-independent neurotoxicity, the anti-apoptotic actions of three structurally distinct PAF antagonists were compared to a panel of plant and fungal benzoic acid derivatives. We found that the PAF antagonist BN 52021 but not FR 49175 or CV 3988 inhibited PAFR-independent neurotoxicity. Orsellinic acid, a fungal-derived benzoic acid, blocked PAF-mediated neuronal apoptosis without affecting PAFR-mediated neuroprotection. These findings demonstrate that PAF can transduce apoptotic death in primary neurons independently of its G-protein coupled receptor, that PAFR activation is neuroprotective, and that orsellinic acid effectively attenuates PAFR-independent neuronal apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fator de Ativação de Plaquetas/toxicidade , Glicoproteínas da Membrana de Plaquetas/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Resorcinóis/farmacologia , Animais , Benzoatos/farmacologia , Inibidores de Caspase , Células Cultivadas , Cruzamentos Genéticos , Inibidores Enzimáticos/farmacologia , Técnicas de Transferência de Genes , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/fisiologia , Fator de Ativação de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
J Neuroinflammation ; 4: 5, 2007 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-17233902

RESUMO

BACKGROUND: Platelet-activating factor (PAF) is implicated in the neuronal damage that accompanies ischemia, prion disease and Alzheimer's disease (AD). Since some epidemiological studies demonstrate that statins, drugs that reduce cholesterol synthesis, have a beneficial effect on mild AD, we examined the effects of two cholesterol synthesis inhibitors on neuronal responses to PAF. METHODS: Primary cortical neurons were treated with cholesterol synthesis inhibitors (simvastatin or squalestatin) prior to incubation with different neurotoxins. The effects of these drugs on neuronal cholesterol levels and neuronal survival were measured. Immunoblots were used to determine the effects of simvastatin or squalestatin on the distribution of the PAF receptor and an enzyme linked immunoassay was used to quantify the amounts of PAF receptor. RESULTS: PAF killed primary neurons in a dose-dependent manner. Pre-treatment with simvastatin or squalestatin reduced neuronal cholesterol and increased the survival of PAF-treated neurons. Neuronal survival was increased 50% by 100 nM simvastatin, or 20 nM squalestatin. The addition of mevalonate restored cholesterol levels, and reversed the protective effect of simvastatin. Simvastatin or squalestatin did not affect the amounts of the PAF receptor but did cause it to disperse from within lipid rafts. CONCLUSION: Treatment of neurons with cholesterol synthesis inhibitors including simvastatin and squalestatin protected neurons against PAF. Treatment caused a percentage of the PAF receptors to disperse from cholesterol-sensitive domains. These results raise the possibility that the effects of statins on neurodegenerative disease are, at least in part, due to desensitisation of neurons to PAF.


Assuntos
Anticolesterolemiantes/farmacologia , Colesterol/biossíntese , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fator de Ativação de Plaquetas/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Camundongos , Neurônios/metabolismo , Fator de Ativação de Plaquetas/antagonistas & inibidores , Fator de Ativação de Plaquetas/farmacologia
13.
Int J Cancer ; 120(6): 1277-83, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17187368

RESUMO

In this study, the roles of matrix metalloproteinase (MMP)-2 and MMP-9 in platelet-activating factor (PAF)-induced experimental pulmonary metastasis of the murine melanoma cell, B16F10, were investigated. An injection of PAF resulted in increases in mRNA expression, protein levels and the activities of both MMP-2 and MMP-9 in the lungs. The overall expression of MMP-9 was stronger than that of MMP-2. The increased MMP-9 expression was inhibited by both NF-kappaB and AP-1 inhibitors, whereas the increased MMP-2 expression was inhibited by only AP-1 inhibitors. Immunohistochemical analysis revealed that MMP-9 was expressed in bronchial epithelial cells as well as in the walls of blood vessels, whereas MMP-2 expression was observed only in bronchial epithelial cells. PAF significantly enhanced the pulmonary metastasis of B16F10, which was inhibited by both NF-kappaB and c-jun inhibitors. MMP-9 inhibitor, but not that of MMP-2, completely inhibited PAF-induced B16F10 metastasis. These data indicate that MMP-9, the expression of which was regulated by NF-kappaB and AP-1, plays a critical role in PAF-induced enhancement of pulmonary melanoma metastasis.


Assuntos
Metaloproteinase 9 da Matriz/fisiologia , Metástase Neoplásica , Fator de Ativação de Plaquetas/toxicidade , Animais , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/análise , Inibidores de Metaloproteinases de Matriz , Melanoma Experimental/induzido quimicamente , Melanoma Experimental/enzimologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , Fator de Ativação de Plaquetas/farmacologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Fator de Transcrição AP-1/antagonistas & inibidores
14.
J Clin Invest ; 115(11): 3185-92, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16276420

RESUMO

Neurologic impairment in HIV-1-associated dementia (HAD) and other neuroinflammatory diseases correlates with injury to dendrites and synapses, but how such injury occurs is not known. We hypothesized that neuroinflammation makes dendrites susceptible to excitotoxic injury following synaptic activity. We report that platelet-activating factor, an inflammatory phospholipid that mediates synaptic plasticity and neurotoxicity and is dramatically elevated in the brain during HAD, promotes dendrite injury following elevated synaptic activity and can replicate HIV-1-associated dendritic pathology. In hippocampal slices exposed to a stable platelet-activating factor analogue, tetanic stimulation that normally induces long-term synaptic potentiation instead promoted development of calcium- and caspase-dependent dendritic beading. Chemical preconditioning with diazoxide, a mitochondrial ATP-sensitive potassium channel agonist, prevented dendritic beading and restored long-term potentiation. In contrast to models invoking excessive glutamate release, these results suggest that physiologic synaptic activity may trigger excitotoxic dendritic injury during chronic neuroinflammation. Furthermore, preconditioning may represent a novel therapeutic strategy for preventing excitotoxic injury while preserving physiologic plasticity.


Assuntos
Espinhas Dendríticas/patologia , Neurônios/patologia , Fator de Ativação de Plaquetas/toxicidade , Transmissão Sináptica/efeitos dos fármacos , Complexo AIDS Demência/metabolismo , Complexo AIDS Demência/patologia , Animais , Cálcio/fisiologia , Caspases/fisiologia , Espinhas Dendríticas/efeitos dos fármacos , Estimulação Elétrica , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Perfusão , Éteres Fosfolipídicos/toxicidade , Fator de Ativação de Plaquetas/agonistas , Fator de Ativação de Plaquetas/análogos & derivados , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
15.
J Immunol ; 175(2): 1192-6, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16002722

RESUMO

C-reactive protein (CRP), an acute phase protein in humans and rabbits, is part of the innate immune system. The role of CRP in host defense has been thought to be largely due to its ability to bind phosphocholine, activate complement, and interact with IgGRs (FcgammaRs). We have shown previously that transgenic rabbit CRP (rbCRP) protects mice from lethal challenges with platelet-activating factor (PAF). To investigate the mechanism of this protection, we created additional lines of transgenic mice that express either wild-type rbCRP, a variant of rbCRP with altered complement activation activity (Y175A), or a variant of rbCRP unable to bind phosphocholine (F66Y/E81K). In the current study, these lines were challenged with a single injection of PAF and their survival monitored. Mice expressing wild-type and Y175A rbCRP were protected against challenge by PAF whereas mice expressing F66Y/E81K rbCRP were not. Treatment with cobra venom factor did not affect survival, confirming the results with the Y175A rbCRP variant and indicating that complement activation was not required to mediate protection. Both wild-type rbCRP and Y175A rbCRP were capable of binding PAF in vitro whereas F66Y/E81K rbCRP was not. Although other interpretations are possible, our results suggest that the protective effect of rbCRP against PAF is due to sequestration of PAF.


Assuntos
Proteína C-Reativa/genética , Proteína C-Reativa/fisiologia , Fosforilcolina/metabolismo , Fator de Ativação de Plaquetas/toxicidade , Animais , Sítios de Ligação/genética , Proteína C-Reativa/biossíntese , Proteína C-Reativa/metabolismo , Ativação do Complemento/fisiologia , Carboidratos da Dieta/administração & dosagem , Proteínas Alimentares/administração & dosagem , Ácido Glutâmico/genética , Lisina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenilalanina/genética , Fator de Ativação de Plaquetas/antagonistas & inibidores , Fator de Ativação de Plaquetas/metabolismo , Ligação Proteica/genética , Coelhos , Análise de Sobrevida , Tirosina/genética
16.
Crit Care Med ; 33(5): 1050-6, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15891335

RESUMO

OBJECTIVE: We reported previously that neuronal nitric oxide synthase (nNOS) is the predominant NOS in rat small intestine and is down-regulated by platelet-activating factor (PAF). The severity of the bowel injury induced by PAF is inversely related to its suppressing effect on nNOS. Here, we investigated whether intestinal perfusion is regulated by nNOS and whether tetrahydrobiopterin, a co-factor and stabilizer of nNOS, reverses PAF-induced intestinal hypoperfusion and injury. SETTING: Animal laboratory. DESIGN: We first examined nNOS regulation of splanchnic blood flow by measuring the perfusion of the heart, lung, ileum, and kidney in rats after a nNOS inhibitor. We then examined the protective effect of tetrahydrobiopterin on PAF-induced bowel injury, mesenteric hypoperfusion, and systemic inflammation. SUBJECTS: Adult male Sprague-Dawley rats. INTERVENTION: In part 1 of the experiment, rats were given 7-nitroindazole (a specific nNOS inhibitor, 50 mg.kg.day). In part 2 of the experiment, rats were treated with tetrahydrobiopterin (20 mg/kg) 5 mins before and 30 mins after PAF challenge (2.2 microg/kg, intravenously) MEASUREMENTS: Perfusion of the heart, lung, ileum, and kidney was measured at 1 and 4 days after 7-nitroindazole, using fluorescent microspheres. Intestinal injury and inflammation (myeloperoxidase content), blood perfusion, calcium dependent-NOS activity, and systemic inflammation (hypotension and hematocrit increase) were assessed 1 hr after PAF with and without tetrahydrobiopterin treatment. RESULTS: In part 1 of the experiment, 7-nitroindazole induced a long-lasting reduction of blood perfusion and inducible NOS expression selectively in the ileum but not in nonsplanchnic organs such as heart, lungs, and kidneys. In part 2, tetrahydrobiopterin protected against PAF-induced intestinal necrosis, hypoperfusion, neutrophil influx, and NOS suppression. It also reversed hypotension and hemoconcentration. Sepiapterin (2 mg/kg, stable tetrahydrobiopterin precursor) also attenuated PAF-induced intestinal injury. CONCLUSIONS: We conclude that nNOS selectively regulates intestinal perfusion. Tetrahydrobiopterin prevents PAF-induced intestinal injury, probably by stabilizing nNOS and maintaining intestinal perfusion.


Assuntos
Antioxidantes/uso terapêutico , Biopterinas/análogos & derivados , Biopterinas/uso terapêutico , Enteropatias/prevenção & controle , Proteínas do Tecido Nervoso/fisiologia , Óxido Nítrico Sintase/fisiologia , Fator de Ativação de Plaquetas/antagonistas & inibidores , Animais , Inibidores Enzimáticos/farmacologia , Indazóis/farmacologia , Enteropatias/etiologia , Masculino , Proteínas do Tecido Nervoso/antagonistas & inibidores , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I , Fator de Ativação de Plaquetas/toxicidade , Pterinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Circulação Esplâncnica/efeitos dos fármacos
17.
Pain ; 111(3): 351-359, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15363879

RESUMO

Platelet-activating factor (PAF) is a potent inflammatory lipid mediator in peripheral tissues. However, its role in mediation of nociception in central nervous system is unknown. In the present study, whether PAF plays some role in pain transduction in the spinal cord was studied in mice. Intrathecal injection of PAF induced tactile pain, tactile allodynia at as low as 10 fg to 1 pg with a peak response at 100 fg, while lyso-PAF was without effect in the range of doses. Tactile allodynia induced by PAF was blocked by a PAF receptor antagonists, TCV-309, WEB 2086 and BN 50739. The expression of PAF receptor mRNA by RT-PCR was observed in DRG and spinal cord in mice. ATP P2X receptor antagonists, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid and 2',3'-O-(2,4,6-trinitrophenyl)adenosine 5-triphosphate, NMDA receptor antagonist, MK 801 and nitric oxide synthetase inhibitor, 7-nitroindazole blocked the PAF-induced tactile allodynia. PAF-induced tactile allodynia and thermal hyperalgesia disappeared in neonatally capsaicin-treated adult mice, while tactile allodynia but not thermal hyperalgesia induced by intrathecally injected alpha,beta-methylene ATP, a P2X receptor agonist, was capsaicin-insensitive. The present study demonstrated that PAF is a potent inducer of tactile allodynia and thermal hyperalgesia at the level of the spinal cord. PAF-evoked tactile allodynia is suggested to be mediated by ATP and the following NMDA and NO cascade through capsaicin-sensitive fiber, different from exogenously injected alpha,beta-methylene ATP which is insensitive to capsaicin treatment.


Assuntos
Hiperalgesia/fisiopatologia , Fator de Ativação de Plaquetas/administração & dosagem , Fator de Ativação de Plaquetas/toxicidade , Animais , Relação Dose-Resposta a Droga , Feminino , Temperatura Alta/efeitos adversos , Hiperalgesia/induzido quimicamente , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos ICR , Medição da Dor/métodos , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Tato/efeitos dos fármacos , Tato/fisiologia
18.
Exp Neurol ; 189(1): 16-24, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15296832

RESUMO

PSD-93, a molecular adaptive protein, binds to and clusters the N-methyl-D-aspartate (NMDA) receptor and assembles a specific set of signaling proteins (for example neuronal nitric oxide synthase, nNOS) around the NMDA receptor at synapses in the central nervous system. This suggests that PSD-93 might mediate many NMDA receptor-dependent physiological and pathophysiological functions. We report here that PSD-93 colocalizes and interacts with the NMDA receptor and neuronal nitric oxide synthase in cultured cortical neurons. Targeted disruption of PSD-93 gene significantly prevented NMDA receptor-nitric oxide signaling-dependent neurotoxicity triggered via platelet-activating factor (PAF) receptor activation. In addition, the deficiency of PSD-93 markedly attenuated platelet-activating factor-induced increase in cyclic guanosine 3',5'-monophosphate (cGMP) and prevented platelet-activating factor-promoted formation of NMDA receptor-neuronal nitric oxide synthase complex. These findings indicate that PSD-93 is involved in the NMDA receptor--nitric oxide-mediated pathological processing of neuronal damage triggered via platelet--activating factor receptor activation. Since platelet-activating factor is a potent neuronal injury mediator during the development of brain trauma, seizures, and ischemia, the present work suggests that PSD-93 might contribute to molecular mechanisms of neuronal damage in these brain disorders.


Assuntos
Córtex Cerebral/citologia , Proteínas do Tecido Nervoso/deficiência , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Fator de Ativação de Plaquetas/toxicidade , Animais , Western Blotting/métodos , Contagem de Células/métodos , Morte Celular/efeitos dos fármacos , Células Cultivadas , GMP Cíclico/metabolismo , Diterpenos/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Imunofluorescência/métodos , Ginkgolídeos , Guanilato Quinases , Peptídeos e Proteínas de Sinalização Intracelular , Lactonas/farmacologia , Proteínas de Membrana , Camundongos , Camundongos Knockout , Microscopia Confocal/métodos , NG-Nitroarginina Metil Éster/farmacologia , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/fisiologia , Neurotoxinas/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Fator de Ativação de Plaquetas/antagonistas & inibidores , Testes de Precipitina/métodos , Propídio , Receptores de N-Metil-D-Aspartato/metabolismo
19.
Neuroreport ; 15(2): 263-6, 2004 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-15076749

RESUMO

Platelet-activating factor (PAF), a bioactive phospholipid implicated in neuronal excitotoxic death, augments the presynaptic release of glutamate. Excessive activation of postsynaptic glutamate receptors and subsequent downstream signals leads to excitotoxicity. The present study proposed that the NMDA receptor/nitric oxide (NO) signal pathway might be involved in PAF-induced neurotoxicity. After the cultured neurons were exposed to PAF for 24 h the percentage of neuronal death increased in a dose-dependent manner. The PAF effects were significantly prevented not only by BN52021, a PAF antagonist, but also by MK-801, an NMDA antagonist, and L-NAME, an NO synthase (NOS) inhibitor. Moreover, the increases in NOS activity and neuronal NOS expression induced by chronic exposure of the cultured neurons to PAF were dramatically blocked by BN52021 and MK-801, respectively. Our findings suggest that the NMDA receptor/NO signaling pathway might contribute to the pathological mechanism of cell death triggered via PAF receptor activation.


Assuntos
Morte Celular/fisiologia , Degeneração Neural/metabolismo , Neurotoxinas/metabolismo , Óxido Nítrico/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Diterpenos/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ginkgolídeos , Lactonas/farmacologia , Camundongos , Degeneração Neural/induzido quimicamente , Degeneração Neural/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotoxinas/toxicidade , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Fator de Ativação de Plaquetas/antagonistas & inibidores , Fator de Ativação de Plaquetas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
20.
Eur Respir J ; 18(1): 122-9, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11510782

RESUMO

Bioactive phospholipids (PL), particularly lysophosphatidylcholine (LPC), are being increasingly implicated in the pathogenesis of various acute and chronic inflammatory disorders, particularly those of the airways, while there is emerging evidence that vitamin E may function as a natural antagonist of these lipid mediators of inflammation. The aims of this study were to document the effects of vitamin E on the inhibition of ciliary beating and damage to structural integrity of human ciliated epithelium induced by the PL, platelet-activating factor (PAF), lyso-PAF and LPC in vitro in relation to the anti-oxidative and membrane-stabilizing properties of the vitamin. Ciliary beat frequency was measured by a phototransistor technique, and damage to structural integrity assessed by a visual-scoring index, while superoxide production by polymorphonuclear leukocytes and membrane-stabilizing potential were measured using lucigenin-enhanced chemiluminescence and haemolytic procedures, respectively. All three PL caused inhibition of ciliary beating and structural damage to human ciliated epithelium by membrane-directed cytotoxic mechanisms, which were potentiated by human polymorphonuclear leukocytes due to induction of oxidant-mediated injury. Both direct and phagocyte-inflicted epithelial injury was attenuated by vitamin E. In haemolytic and chemiluminescence assays, vitamin E neutralized both the membrane-destabilizing and pro-oxidative actions of all three PL, while spectrophotometric analysis of mixtures of vitamin E with PAF, lyso-PAF and LPC revealed alterations in peak intensity, as well as peak shifts, indicative of physicochemical interactions between the vitamin and the PL. Vitamin E status may be a determinant of susceptibility to phospholipid-mediated airway inflammation and damage.


Assuntos
Depuração Mucociliar/efeitos dos fármacos , Mucosa Nasal/efeitos dos fármacos , Fosfolipídeos/antagonistas & inibidores , Fator de Ativação de Plaquetas/análogos & derivados , Vitamina E/farmacologia , Células Cultivadas , Humanos , Lisofosfatidilcolinas/antagonistas & inibidores , Lisofosfatidilcolinas/toxicidade , Fosfolipídeos/toxicidade , Fator de Ativação de Plaquetas/antagonistas & inibidores , Fator de Ativação de Plaquetas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...